101
|
Structure and biological properties of five Pt(II) complexes as potential anticancer agents. J Inorg Biochem 2018; 185:10-16. [PMID: 29730232 DOI: 10.1016/j.jinorgbio.2018.04.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/22/2018] [Accepted: 04/22/2018] [Indexed: 10/17/2022]
Abstract
We synthesized and validated five Schiff base Pt(II) complexes derived from 2-hydroxy-1-naphthaldehyde benzoyl hydrazone and its derivatives, which are modified at the benzohydrazide structures (L1-L5). The complexes were [Pt(L1)(DMSO)Cl] (C1), [Pt(L2)(DMSO)Cl] (C2), [Pt(L3)(DMSO)Cl] (C3), [Pt(L4)(DMSO)Cl] (C4), and [Pt(L5)(DMSO)Cl] (C5). Crystal structures showed that the Pt centers of all complexes were tetra-coordinated with other atoms. The structure-activity relationships and anticancer mechanisms of the complexes were explored. These five Pt(II) complexes were toxic at micromolar doses and exhibited cytotoxicity similar to or somewhat higher than that of cisplatin, with IC50 values ranging from 4.38 μM to 25.16 μM. The complexes exerted chemotherapeutic effects via inhibition of telomerase by targeting the c-myc promoter and down-regulating the expression of human telomerase reverse transcriptase, consequently triggering cell apoptosis. In addition, Pt(II) complexes also caused cell cycle arrest at S-phase, leading to the down-regulation of cdc25 A, cyclin A2, and CDK2 and up-regulation of p53, p27, and p21 proteins. Other complex-associated events were reactive oxygen species production, transformation of the mitochondrial membrane potential (Δψm), release of cytochrome c, regulation of Bcl-2 family protein expression, facilitated release of apoptotic active substances, and activation of caspases to induce apoptosis.
Collapse
|
102
|
Park HR, Kim YW, Park JH, Maeng YH, Nojima T, Hashimoto H, Park YK. Low Expression of P63 and P73 in Osteosarcoma. TUMORI JOURNAL 2018; 90:239-43. [PMID: 15237589 DOI: 10.1177/030089160409000214] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background The recent discovery of two p53-related genes, p63 and p73, has revealed an additional level of complexity to the study of p53 function. Both genes encode multiple proteins arising from alternative promoter usage and splicing, with transactivation, DNA-binding, and tetramerization domains. Recent data support a role for p63 in squamous and transitional cell carcinomas, as well as in certain lymphomas and thymomas. Methods To characterize the involvement of p63 and p73 in the development of osteosarcoma, we analyzed the expression and mutation of TAp63 and TAp73 in six osteosarcoma cell lines and twelve osteosarcoma specimens. Results Semiquantitative DNA/PCR analysis revealed that eight (67%) and six (50%) out of twelve osteosarcoma specimens showed significantly reduced levels of p63 and p73 transcription, respectively. Direct sequencing of the entire coding region detected no mutations in cell lines or osteosarcoma specimens. Conclusions Our data suggest that low expression of p63 and p73 is relatively common in osteosarcomas and might contribute to their molecular pathogenesis.
Collapse
Affiliation(s)
- Hye-Rim Park
- Department of Pathology, College of Medicine, Hallym University, Anyang, Korea
| | | | | | | | | | | | | |
Collapse
|
103
|
Targeting p53 as a promising therapeutic option for cancer by re-activating the wt or mutant p53’s tumor suppression. Future Med Chem 2018; 10:755-777. [DOI: 10.4155/fmc-2017-0175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
p53 protein, a product of the TP53 tumor suppressor gene, controls the cellular genome’s integrity and is an important regulator of cell cycling, proliferation, apoptosis and metabolism. Mutations of TP53 or inactivation of its gene product are among the first events initiating malignant transformation. The consequent loss of control over the cell cycle, resulting in accelerated cell proliferation and facilitating metabolic reprogramming, gives the initiated (premalignant) cells numerous advantages over healthy cells. Interestingly, p53 status is not only an important marker in cancer diagnosis; it has also become a promising target of personalized therapy. Depending on the TP53 status different therapeutic options have been developed. (Re)-activation of p53 functionality in cancer cells offers promising new alternatives to existing oncological therapies.
Collapse
|
104
|
Meyer G, González-Gómez M. The heterogeneity of human Cajal-Retzius neurons. Semin Cell Dev Biol 2018; 76:101-111. [DOI: 10.1016/j.semcdb.2017.08.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 12/29/2022]
|
105
|
Lee T, Pelletier J. Dependence of p53-deficient cells on the DHX9 DExH-box helicase. Oncotarget 2018; 8:30908-30921. [PMID: 28427210 PMCID: PMC5458177 DOI: 10.18632/oncotarget.15889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/21/2017] [Indexed: 12/13/2022] Open
Abstract
DHX9 is a DExH-box helicase family member with key regulatory roles in a broad range of cellular processes. It participates at multiple levels of gene regulation, including DNA replication, transcription, translation, RNA transport, and microRNA processing. It has been implicated in tumorigenesis and recent evidence suggests that it may be a promising chemotherapeutic target. Previous studies have determined that DHX9 suppression elicits an apoptotic or senescence response by activating p53 signaling. Here, we show that DHX9 inhibition can also have deleterious effects in cells lacking functional p53. Loss of DHX9 led to increased cell death in p53-deficient mouse lymphomas and HCT116 human colon cancer cells, and G0/G1 cell cycle arrest in p53-deficient mouse embryonic fibroblasts. Analysis of mRNA levels for p53 transcriptional targets showed that a subset of p53 targets in the p53-null lymphomas and HCT116 cells were activated despite the absence of functional p53. This implies an alternative pathway of DHX9-mediated activation of cell death and cell cycle arrest in p53-deficient cells and supports the feasibility of targeting DHX9 in p53-deficient tumors.
Collapse
Affiliation(s)
- Teresa Lee
- Department of Biochemistry, McGill University, Montreal, Quebec, H3G 1Y6, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec, H3G 1Y6, Canada.,Department of Oncology, McGill University, Montreal, Quebec, H3G 1Y6, Canada.,Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Quebec, H3G 1Y6, Canada
| |
Collapse
|
106
|
Relationship of p73 gene polymorphism and additional gene-smoking and gene-obesity interaction with non-small cell lung cancer risk. Oncotarget 2018; 8:34423-34428. [PMID: 28415779 PMCID: PMC5470979 DOI: 10.18632/oncotarget.16257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 03/01/2017] [Indexed: 11/25/2022] Open
Abstract
Aim The aim of this study was to investigate the impact of G4C14-to-A4T14 polymorphism within P73 gene and additional interactions with current smoking and obesity on non-small cell lung cancer (NSCLC) risk in a Chinese population. Results Logistic regression analysis showed a significant association between genotypes of the AT allele in G4C14-to-A4T14 and decreased NSCLC risk. NSCLC risk was significantly lower in carriers of the G4C14-to-A4T14- AT allele than those with GC/GC genotype (AT/AT + GC/AT versus GC/GC), adjusted OR (95%CI) = 0.68 (0.55–0.93). We also found that the OR (95%CI) was 1.88 (1.32-2.47) for current smokers compared with never smokers and 0.69 (0.40–0.95) for obese subjects compared to participants with normal BMI. Never smokers with AT/AT or GC/AT of the G4C14-to-A4T14 genotype have the lowest NSCLC risk compared with smokers with the GC/GC genotype after covariates adjustment, OR (95%CI) = 0.52 (0.26-0.87). Obese participants with G4C14-to-A4T14- AT/AT or GC/AT genotype have the lowest NSCLC risk compared with non- obese subjects with the GC/GC genotype after adjusting for covariates, OR (95% CI) = 0.56 (0.33–0.85). Materials and Methods A logistic regression model was used to examine the association between G4C14-to-A4T14 polymorphism and NSCLC, and its interaction with current smoking and obesity. The odds ratios (OR) and 95% confident intervals (95%CI) were calculated. Conclusions Our results support an important association between the G4C14-to-A4T14 and decreased NSCLC risk and additional impact of an interaction between G4C14-to-A4T14 and smoking or obesity on NSCLC risk.
Collapse
|
107
|
Ozaki T, Yu M, Yin D, Sun D, Zhu Y, Bu Y, Sang M. Impact of RUNX2 on drug-resistant human pancreatic cancer cells with p53 mutations. BMC Cancer 2018; 18:309. [PMID: 29558908 PMCID: PMC5861661 DOI: 10.1186/s12885-018-4217-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 03/12/2018] [Indexed: 12/20/2022] Open
Abstract
Background Despite the remarkable advances in the early diagnosis and treatment, overall 5-year survival rate of patients with pancreatic cancer is less than 10%. Gemcitabine (GEM), a cytidine nucleoside analogue and ribonucleotide reductase inhibitor, is a primary option for patients with advanced pancreatic cancer; however, its clinical efficacy is extremely limited. This unfavorable clinical outcome of pancreatic cancer patients is at least in part attributable to their poor response to anti-cancer drugs such as GEM. Thus, it is urgent to understand the precise molecular basis behind the drug-resistant property of pancreatic cancer and also to develop a novel strategy to overcome this deadly disease. Review Accumulating evidence strongly suggests that p53 mutations contribute to the acquisition and/or maintenance of drug-resistant property of pancreatic cancer. Indeed, certain p53 mutants render pancreatic cancer cells much more resistant to GEM, implying that p53 mutation is one of the critical determinants of GEM sensitivity. Intriguingly, runt-related transcription factor 2 (RUNX2) is expressed at higher level in numerous human cancers such as pancreatic cancer and osteosarcoma, indicating that, in addition to its pro-osteogenic role, RUNX2 has a pro-oncogenic potential. Moreover, a growing body of evidence implies that a variety of miRNAs suppress malignant phenotypes of pancreatic cancer cells including drug resistance through the down-regulation of RUNX2. Recently, we have found for the first time that forced depletion of RUNX2 significantly increases GEM sensitivity of p53-null as well as p53-mutated pancreatic cancer cells through the stimulation of p53 family TAp63/TAp73-dependent cell death pathway. Conclusions Together, it is likely that RUNX2 is one of the promising molecular targets for the treatment of the patients with pancreatic cancer regardless of their p53 status. In this review article, we will discuss how to overcome the serious drug-resistant phenotype of pancreatic cancer.
Collapse
Affiliation(s)
- Toshinori Ozaki
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, 260-8717, Japan.
| | - Meng Yu
- Department of Laboratory Animal of China Medical University, Shenyang, 110001, People's Republic of China
| | - Danjing Yin
- Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, People's Republic of China
| | - Dan Sun
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yuyan Zhu
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Youquan Bu
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Meixiang Sang
- Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, People's Republic of China
| |
Collapse
|
108
|
Najem S, Langemann D, Appl B, Trochimiuk M, Hundsdoerfer P, Reinshagen K, Eschenburg G. Smac mimetic LCL161 supports neuroblastoma chemotherapy in a drug class-dependent manner and synergistically interacts with ALK inhibitor TAE684 in cells with ALK mutation F1174L. Oncotarget 2018; 7:72634-72653. [PMID: 27655666 PMCID: PMC5341933 DOI: 10.18632/oncotarget.12055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/27/2016] [Indexed: 12/21/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor during infancy and childhood. Outcome of high-risk and late-stage disease remains poor despite intensive treatment regimens. Suppressing inhibitor of apoptosis proteins (IAPs) using Smac mimetics (SM) significantly sensitizes neuroblastoma (NB) cells for chemotherapy, however strongly dependent on the cytotoxic drug combined with SM. Therefore, a systematic analysis of the impact of SM in combination with different classes of chemotherapeutics was of crucial importance. Treatment of NB cell lines with SM LCL161 and vinca alkaloids revealed a strong synergistic inhibition of proliferation and significant induction of apoptosis in virtually all established and de novo NB cell lines (n=8). In contrast, combination of anthracyclines or topoisomerase inhibitors with LCL161 showed a synergism for single drugs and/or cell lines only. Furthermore, we could show that insensibility to LCL161-mediated sensitization for chemotherapeutics is associated with aberrant activation of anaplastic lymphoma kinase (ALK) by common mutation F1174L. Inhibition of ALK using TAE684 is able to overcome this resistance in a synergistic fashion, a finding that could be highly relevant for improvement of neuroblastoma therapy.
Collapse
Affiliation(s)
- Safiullah Najem
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doerte Langemann
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birgit Appl
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Magdalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Hundsdoerfer
- Department of Pediatric Oncology/Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Georg Eschenburg
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
109
|
Abstract
Glucose is the key source for most organisms to provide energy, as well as the key source for metabolites to generate building blocks in cells. The deregulation of glucose homeostasis occurs in various diseases, including the enhanced aerobic glycolysis that is observed in cancers, and insulin resistance in diabetes. Although p53 is thought to suppress tumorigenesis primarily by inducing cell cycle arrest, apoptosis, and senescence in response to stress, the non-canonical functions of p53 in cellular energy homeostasis and metabolism are also emerging as critical factors for tumor suppression. Increasing evidence suggests that p53 plays a significant role in regulating glucose homeostasis. Furthermore, the p53 family members p63 and p73, as well as gain-of-function p53 mutants, are also involved in glucose metabolism. Indeed, how this protein family regulates cellular energy levels is complicated and difficult to disentangle. This review discusses the roles of the p53 family in multiple metabolic processes, such as glycolysis, gluconeogenesis, aerobic respiration, and autophagy. We also discuss how the dysregulation of the p53 family in these processes leads to diseases such as cancer and diabetes. Elucidating the complexities of the p53 family members in glucose homeostasis will improve our understanding of these diseases.
Collapse
|
110
|
p73 coordinates with Δ133p53 to promote DNA double-strand break repair. Cell Death Differ 2018; 25:1063-1079. [PMID: 29511339 PMCID: PMC5988805 DOI: 10.1038/s41418-018-0085-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/18/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022] Open
Abstract
Tumour repressor p53 isoform Δ133p53 is a target gene of p53 and an antagonist of p53-mediated apoptotic activity. We recently demonstrated that Δ133p53 promotes DNA double-strand break (DSB) repair by upregulating transcription of the repair genes RAD51, LIG4 and RAD52 in a p53-independent manner. However, Δ133p53 lacks the transactivation domain of full-length p53, and the mechanism by which it exerts transcriptional activity independently of full-length p53 remains unclear. In this report, we describe the accumulation of high levels of both Δ133p53 and p73 (a p53 family member) at 24 h post γ-irradiation (hpi). Δ133p53 can form a complex with p73 upon γ-irradiation. The co-expression of Δ133p53 and p73, but not either protein alone, can significantly promote DNA DSB repair mechanisms, including homologous recombination (HR), non-homologous end joining (NHEJ) and single-strand annealing (SSA). p73 and Δ133p53 act synergistically to promote the expression of RAD51, LIG4 and RAD52 by joining together to bind to region containing a Δ133p53-responsive element (RE) and a p73-RE in the promoters of all three repair genes. In addition to its accumulation at 24 hpi, p73 protein expression also peaks at 4 hpi. The depletion of p73 not only reduces early-stage apoptotic frequency (4–6 hpi), but also significantly increases later-stage DNA DSB accumulation (48 hpi), leading to cell cycle arrest in the G2 phase and, ultimately, cell senescence. In summary, the apoptotic regulator p73 also coordinates with Δ133p53 to promote DNA DSB repair, and the loss of function of p73 in DNA DSB repair may underlie spontaneous and carcinogen-induced tumorigenesis in p73 knockout mice.
Collapse
|
111
|
Ozaki T, Nakamura M, Ogata T, Sang M, Yoda H, Hiraoka K, Sang M, Shimozato O. Depletion of pro-oncogenic RUNX2 enhances gemcitabine (GEM) sensitivity of p53-mutated pancreatic cancer Panc-1 cells through the induction of pro-apoptotic TAp63. Oncotarget 2018; 7:71937-71950. [PMID: 27713122 PMCID: PMC5342134 DOI: 10.18632/oncotarget.12433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/25/2016] [Indexed: 12/13/2022] Open
Abstract
Recently, we have described that siRNA-mediated silencing of runt-related transcription factor 2 (RUNX2) improves anti-cancer drug gemcitabine (GEM) sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the augmentation of p53 family TAp63-dependent cell death pathway. In this manuscript, we have extended our study to p53-mutated human pancreatic cancer Panc-1 cells. According to our present results, knockdown of mutant p53 alone had a marginal effect on GEM-mediated cell death of Panc-1 cells. We then sought to deplete RUNX2 using siRNA in Panc-1 cells and examined its effect on GEM sensitivity. Under our experimental conditions, RUNX2 knockdown caused a significant enhancement of GEM sensitivity of Panc-1 cells. Notably, GEM-mediated induction of TAp63 but not of TAp73 was further stimulated in RUNX2-depleted Panc-1 cells, indicating that, like AsPC-1 cells, TAp63 might play a pivotal role in the regulation of GEM sensitivity of Panc-1 cells. Consistent with this notion, forced expression of TAp63α in Panc-1 cells promoted cell cycle arrest and/or cell death, and massively increased luciferase activities driven by TAp63-target gene promoters such as p21WAF1 and NOXA. In addition, immunoprecipitation experiments indicated that RUNX2 forms a complex with TAp63 in Panc-1 cells. Taken together, our current observations strongly suggest that depletion of RUNX2 enhances the cytotoxic effect of GEM on p53-mutated Panc-1 cells through the stimulation of TAp63-dependent cell death pathway even in the presence of a large amount of pro-oncogenic mutant p53, and might provide an attractive strategy to treat pancreatic cancer patients with p53 mutations.
Collapse
Affiliation(s)
- Toshinori Ozaki
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Mizuyo Nakamura
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Takehiro Ogata
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Meijie Sang
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, Japan.,Department of Regenerative Medicine, Graduate School of Medicine and Pharmatheutical Science, University of Toyama, Toyama, Japan
| | - Hiroyuki Yoda
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Kiriko Hiraoka
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Meixiang Sang
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, Japan.,Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Osamu Shimozato
- Laboratory of DNA Damage Signaling, Chiba Cancer Center Research Institute, Chiba, Japan
| |
Collapse
|
112
|
Bhaumik P, Ghosh P, Biswas A, Ghosh S, Pal S, Sarkar B, Kumar Dey S. Rare Intronic Variations inTP73Gene Found in Patients with Alzheimer’sDisease. INT J HUM GENET 2018. [DOI: 10.1080/09723757.2017.1421438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Pranami Bhaumik
- Department of Biotechnology, School of Biotechnology and Biological Sciences, Maulana Abul Kalam Azad University of Technology, West Bengal (Formerly known as West Bengal University of Technology) BF – 142, Salt Lake City, Sector I. Kolkata 700 064, West Bengal, India
| | - Priyanka Ghosh
- Department of Biotechnology, School of Biotechnology and Biological Sciences, Maulana Abul Kalam Azad University of Technology, West Bengal (Formerly known as West Bengal University of Technology) BF – 142, Salt Lake City, Sector I. Kolkata 700 064, West Bengal, India
| | - Atanu Biswas
- Department of Neurology, Bangur Institute of Neurosciences, 52/1A, S.N. Pandit Street, Kolkata 700 025, West Bengal, India
| | - Sujay Ghosh
- Department of Zoology, University of Calcutta, (Ballygunge Science College Campus), 35 Ballygunge Circular Road., Kolkata 700 019, West Bengal, India
| | - Sandip Pal
- Department of Neurology, Burdwan Medical College, Burdwan 713 104, West Bengal, India
| | - Biswanath Sarkar
- DNA Laboratory, Anthropological Survey of India, 27 Jawaharlal Nehru Road Kolkata 700 016, West Bengal, India
| | - Subrata Kumar Dey
- Department of Biotechnology, School of Biotechnology and Biological Sciences, Maulana Abul Kalam Azad University of Technology, West Bengal (Formerly known as West Bengal University of Technology) BF – 142, Salt Lake City, Sector I. Kolkata 700 064, West Bengal, India
| |
Collapse
|
113
|
Wang B, Liu X, Liu H, Guo J, Zhang T, Zhou N, Ma Y, Yu H, Chen L, Ren Z, Fan K, Tian X. Differential expressions of MDM2 and TAP73 in cancer and cancer-adjacent tissues in patients with non-small-cell lung carcinoma. Pulmonology 2018; 24:S2173-5115(17)30153-7. [PMID: 29452959 DOI: 10.1016/j.rppnen.2017.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 08/04/2017] [Accepted: 08/08/2017] [Indexed: 01/15/2023] Open
Abstract
AIM To investigate the differences in mRNA and protein expressions of MDM2 (mouse double minute 2 homolog) and P73 in cancer and cancer-adjacent tissues in patients with non-small-cell lung carcinoma (NSCLC). MATERIALS AND METHODS We compared the protein expressions of MDM2 and P73 in lung cancer and cancer-adjacent tissues in NSCLC patients by IHC (immunohistochemistry) and WB (Western blot). We divided the NSCLC patients into two subgroups, adenocarcinoma and squamous carcinoma. The mRNA expressions of two main isoforms of P73, TAP73 and DNP73, as well as the ratio of DNP73/TAP73 were analyzed by qPCR (quantitative real-time PCR) in the two tissues in all NSCLC patients and in patients with adenocarcinoma or squamous carcinoma, respectively. RESULTS WB results did not show significant differences in MDM2 and P73 protein expressions in lung cancer and cancer-adjacent tissues. However, IHC results indicated that MDM2 expression significantly increased in cancer tissues in female patients, but not male patients. In addition, TAP73 mRNA expression significantly increased in cancer tissues in all NSCLC patients (p=0.002) and in patients with adenocarcinoma (p=0.01); while there was no significant difference in DNP73 mRNA expression. Hence the fold-change of DNP73/TAP73 ratio significantly decreased (p=0.0003) in cancer tissues in all NSCLC patients and in patients with adenocarcinoma. CONCLUSIONS TAP73 mRNA expression significantly increased in cancer tissues than cancer-adjacent tissues in all NSCLC patients and in patients with adenocarcinoma. Meanwhile, the fold-change of DNP73/TAP73 ratio was similar to TAP73. MDM2 protein expression significantly increased in cancer tissues in female NSCLC patients.
Collapse
Affiliation(s)
- B Wang
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - X Liu
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - H Liu
- Department of Thoracic Surgery, PLA 309 Hospital, Beijing, China
| | - J Guo
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - T Zhang
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - N Zhou
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - Y Ma
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - H Yu
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - L Chen
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - Z Ren
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - K Fan
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China
| | - X Tian
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
114
|
Billant O, Léon A, Le Guellec S, Friocourt G, Blondel M, Voisset C. The dominant-negative interplay between p53, p63 and p73: A family affair. Oncotarget 2018; 7:69549-69564. [PMID: 27589690 PMCID: PMC5342497 DOI: 10.18632/oncotarget.11774] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 07/10/2016] [Indexed: 12/15/2022] Open
Abstract
The tumor suppression activity of p53 is frequently impaired in cancers even when a wild-type copy of the gene is still present, suggesting that a dominant-negative effect is exerted by some of p53 mutants and isoforms. p63 and p73, which are related to p53, have also been reported to be subjected to a similar loss of function, suggesting that a dominant-negative interplay might happen between p53, p63 and p73. However, to which extent p53 hotspot mutants and isoforms of p53, p63 and p73 are able to interfere with the tumor suppressive activity of their siblings as well as the underlying mechanisms remain undeciphered. Using yeast, we showed that a dominant-negative effect is widely spread within the p53/p63/p73 family as all p53 loss-of-function hotspot mutants and several of the isoforms of p53 and p73 tested exhibit a dominant-negative potential. In addition, we found that this dominant-negative effect over p53 wild-type is based on tetramer poisoning through the formation of inactive hetero-tetramers and does not rely on a prion-like mechanism contrary to what has been previously suggested. We also showed that mutant p53-R175H gains the ability to inhibit p63 and p73 activity by a mechanism that is only partially based on tetramerization.
Collapse
Affiliation(s)
- Olivier Billant
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Alice Léon
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Solenn Le Guellec
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Gaëlle Friocourt
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Marc Blondel
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Cécile Voisset
- Inserm UMR 1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé, Etablissement Français du Sang (EFS) Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| |
Collapse
|
115
|
Protein phosphatase 2ACα gene knock-out results in cortical atrophy through activating hippo cascade in neuronal progenitor cells. Int J Biochem Cell Biol 2018; 95:53-62. [DOI: 10.1016/j.biocel.2017.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/02/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022]
|
116
|
Kim DW, Kim KC, Kim KB, Dunn CT, Park KS. Transcriptional deregulation underlying the pathogenesis of small cell lung cancer. Transl Lung Cancer Res 2018. [PMID: 29535909 DOI: 10.21037/tlcr.2017.10.07] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The discovery of recurrent alterations in genes encoding transcription regulators and chromatin modifiers is one of the most important recent developments in the study of the small cell lung cancer (SCLC) genome. With advances in models and analytical methods, the field of SCLC biology has seen remarkable progress in understanding the deregulated transcription networks linked to the tumor development and malignant progression. This review will discuss recent discoveries on the roles of RB and P53 family of tumor suppressors and MYC family of oncogenes in tumor initiation and development. It will also describe the roles of lineage-specific factors in neuroendocrine (NE) cell differentiation and homeostasis and the roles of epigenetic alterations driven by changes in NFIB and chromatin modifiers in malignant progression and chemoresistance. These recent findings have led to a model of transcriptional network in which multiple pathways converge on regulatory regions of crucial genes linked to tumor development. Validation of this model and characterization of target genes will provide critical insights into the biology of SCLC and novel strategies for tumor intervention.
Collapse
Affiliation(s)
- Dong-Wook Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Keun-Cheol Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA.,Department of Biological Sciences, Kangwon National University, Chuncheon, Korea
| | - Kee-Beom Kim
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Colin T Dunn
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Kwon-Sik Park
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
117
|
Sasaki Y, Tamura M, Takeda K, Ogi K, Nakagaki T, Koyama R, Idogawa M, Hiratsuka H, Tokino T. Identification and characterization of the intercellular adhesion molecule-2 gene as a novel p53 target. Oncotarget 2018; 7:61426-61437. [PMID: 27556181 PMCID: PMC5308662 DOI: 10.18632/oncotarget.11366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/08/2016] [Indexed: 12/19/2022] Open
Abstract
The p53 tumor suppressor inhibits cell growth through the activation of both cell cycle arrest and apoptosis, which maintain genome stability and prevent cancer development. Here, we report that intercellular adhesion molecule-2 (ICAM2) is transcriptionally activated by p53. Specifically, ICAM2 is induced by the p53 family and DNA damage in a p53-dependent manner. We identified a p53 binding sequence located within the ICAM2 gene that is responsive to wild-type p53, TAp73, and TAp63. In terms of function, we found that the ectopic expression of ICAM2 inhibited cancer cell migration and invasion. In addition, we demonstrated that silencing endogenous ICAM2 in cancer cells caused a marked increase in extracellular signal-regulated kinase (ERK) phosphorylation levels, suggesting that ICAM2 inhibits migration and invasion of cancer cells by suppressing ERK signaling. Moreover, ICAM2 is underexpressed in human cancer tissues containing mutant p53 as compared to those with wild-type p53. Notably, the decreased expression of ICAM2 is associated with poor survival in patients with various cancers. Our findings demonstrate that ICAM2 induction by p53 has a key role in inhibiting migration and invasion.
Collapse
Affiliation(s)
- Yasushi Sasaki
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Miyuki Tamura
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Kousuke Takeda
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan.,Department of Oral Surgery, Sapporo Medical University, Sapporo, Japan
| | - Kazuhiro Ogi
- Department of Oral Surgery, Sapporo Medical University, Sapporo, Japan
| | - Takafumi Nakagaki
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan.,Department of Oral Surgery, Sapporo Medical University, Sapporo, Japan
| | - Ryota Koyama
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Masashi Idogawa
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | | | - Takashi Tokino
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
118
|
Molina-Pinelo S, Salinas A, Moreno-Mata N, Ferrer I, Suarez R, Andrés-León E, Rodríguez-Paredes M, Gutekunst J, Jantus-Lewintre E, Camps C, Carnero A, Paz-Ares L. Impact of DLK1-DIO3 imprinted cluster hypomethylation in smoker patients with lung cancer. Oncotarget 2018; 9:4395-4410. [PMID: 29435111 PMCID: PMC5796982 DOI: 10.18632/oncotarget.10611] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 06/02/2016] [Indexed: 12/21/2022] Open
Abstract
DNA methylation is important for gene expression and genome stability, and its disruption is thought to play a key role in the initiation and progression of cancer and other diseases. The DLK1-DIO3 cluster has been shown to be imprinted in humans, and some of its components are relevant to diverse pathological processes. The purpose of this study was to assess the methylation patterns of the DLK1-DIO3 cluster in patients with lung cancer to study its relevance in the pathogenesis of this disease. We found a characteristic methylation pattern of this cluster in smoking associated lung cancer, as compared to normal lung tissue. This methylation profile is not patent however in lung cancer of never smokers nor in lung tissue of COPD patients. We found 3 deregulated protein-coding genes at this locus: one was hypermethylated (DIO3) and two were hypomethylated (DLK1 and RTL1). Statistically significant differences were also detected in two different families of SNORDs, two miRNA clusters and four lncRNAs (MEG3, MEG8, MEG9 and LINC00524). These findings were validated using data from the cancer genome atlas (TCGA) database. We have then showed an inverse correlation between DNA methylation and expression levels in 5 randomly selected genes. Several targets of miRNAs included in the DLK1-DIO3 cluster have been experimentally verified as tumor suppressors. All of these results suggest that the dysmethylation of the imprinted DLK1-DIO3 cluster could have a relevant role in the pathogenesis of lung cancer in current and former smokers and may be used for diagnostic and/or therapeutic purposes.
Collapse
Affiliation(s)
- Sonia Molina-Pinelo
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
- Medical Oncology Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- CIBER de Cáncer, Madrid, Spain
| | - Ana Salinas
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - Nicolás Moreno-Mata
- Thoracic Surgery Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Irene Ferrer
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
- Medical Oncology Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- CIBER de Cáncer, Madrid, Spain
| | - Rocío Suarez
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
- Medical Oncology Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- CIBER de Cáncer, Madrid, Spain
| | - Eduardo Andrés-León
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - Manuel Rodríguez-Paredes
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany
- University Tumor Center Düsseldorf, University of Düsseldorf, Düsseldorf, Germany
| | - Julian Gutekunst
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación para la Investigación del Hospital General Universitario de Valencia, Valencia, Spain
- Department of Biotechnology, Universitat Politècnica de Valencia, Valencia, Spain
| | - Carlos Camps
- Department of Medicine, University of Valencia, Valencia, Spain
- Department of Medical Oncology, Hospital General Universitario de Valencia, Valencia, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
- CIBER de Cáncer, Madrid, Spain
| | - Luis Paz-Ares
- Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
- Medical Oncology Department, Hospital Universitario Doce de Octubre & Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- CIBER de Cáncer, Madrid, Spain
| |
Collapse
|
119
|
Abstract
microRNAs (miRNAs) have vital roles in regulating gene expression-contributing to major diseases like cancer and heart disease. Over the last decade, thousands of miRNAs have been discovered through high throughput sequencing-based annotation. Different classes have been described, as well as a great dynamic range of expression levels. While sequencing approaches provide insight into biogenesis and allow confident identification, there is a need for additional methods for validation and characterization. Northern blotting was one of the first techniques used for studying miRNAs, and remains one of the most valuable as it avoids enzymatic manipulation of miRNA transcripts. Blotting can also provide insight into biogenesis by revealing RNA processing intermediates. Compared to sequencing, however, northern blotting is a relatively insensitive technology. This creates a challenge for detecting low expressed miRNAs, particularly those produced by inefficient, non-canonical pathways. In this chapter, we describe a strategy to study such miRNAs by northern blotting that involves ectopic expression of both miRNAs and miRNA-binding Argonaute (Ago) proteins. Through use of epitope tags, this strategy also provides a convenient method for verification of small RNA competency to be loaded into regulatory complexes.
Collapse
|
120
|
Affiliation(s)
- Francesca Pentimalli
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori – IRCCS -Fondazione G, Pascale, Naples, Italy
| |
Collapse
|
121
|
Søberg K, Skålhegg BS. The Molecular Basis for Specificity at the Level of the Protein Kinase a Catalytic Subunit. Front Endocrinol (Lausanne) 2018; 9:538. [PMID: 30258407 PMCID: PMC6143667 DOI: 10.3389/fendo.2018.00538] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022] Open
Abstract
Assembly of multi enzyme complexes at subcellular localizations by anchoring- and scaffolding proteins represents a pivotal mechanism for achieving spatiotemporal regulation of cellular signaling after hormone receptor targeting [for review, see (1)]. In the 3' 5'-cyclic adenosine monophosphate (cAMP) dependent protein kinase (PKA) signaling pathway it is generally accepted that specificity is secured at several levels. This includes at the first level stimulation of receptors coupled to heterotrimeric G proteins which through stimulation of adenylyl cyclase (AC) forms the second messenger cAMP. Cyclic AMP has several receptors including PKA. PKA is a tetrameric holoenzyme consisting of a regulatory (R) subunit dimer and two catalytic (C) subunits. The R subunit is the receptor for cAMP and compartmentalizes cAMP signals through binding to cell and tissue-specifically expressed A kinase anchoring proteins (AKAPs). The current dogma tells that in the presence of cAMP, PKA dissociates into an R subunit dimer and two C subunits which are free to phosphorylate relevant substrates in the cytosol and nucleus. The release of the C subunit has raised the question how specificity of the cAMP and PKA signaling pathway is maintained when the C subunit no longer is attached to the R subunit-AKAP complex. An increasing body of evidence points toward a regulatory role of the cAMP and PKA signaling pathway by targeting the C subunits to various C subunit binding proteins in the cytosol and nucleus. Moreover, recent identification of isoform specific amino acid sequences, motifs and three dimensional structures have together provided new insight into how PKA at the level of the C subunit may act in a highly isoform-specific fashion. Here we discuss recent understanding of specificity of the cAMP and PKA signaling pathway based on C subunit subcellular targeting as well as evolution of the C subunit structure that may contribute to the dynamic regulation of C subunit activity.
Collapse
Affiliation(s)
- Kristoffer Søberg
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Bjørn Steen Skålhegg
- Section for Molecular Nutrition, University of Oslo, Oslo, Norway
- *Correspondence: Bjørn Steen Skålhegg
| |
Collapse
|
122
|
Agostini M, Annicchiarico-Petruzzelli M, Melino G, Rufini A. Metabolic pathways regulated by TAp73 in response to oxidative stress. Oncotarget 2017; 7:29881-900. [PMID: 27119504 PMCID: PMC5058650 DOI: 10.18632/oncotarget.8935] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/16/2016] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species are involved in both physiological and pathological processes including neurodegeneration and cancer. Therefore, cells have developed scavenging mechanisms to maintain redox homeostasis under control. Tumor suppressor genes play a critical role in the regulation of antioxidant genes. Here, we investigated whether the tumor suppressor gene TAp73 is involved in the regulation of metabolic adaptations triggered in response to oxidative stress. H2O2 treatment resulted in numerous biochemical changes in both control and TAp73 knockout (TAp73−/−) mouse embryonic fibroblasts, however the extent of these changes was more pronounced in TAp73−/− cells when compared to control cells. In particular, loss of TAp73 led to alterations in glucose, nucleotide and amino acid metabolism. In addition, H2O2 treatment resulted in increased pentose phosphate pathway (PPP) activity in null mouse embryonic fibroblasts. Overall, our results suggest that in the absence of TAp73, H2O2 treatment results in an enhanced oxidative environment, and at the same time in an increased pro-anabolic phenotype. In conclusion, the metabolic profile observed reinforces the role of TAp73 as tumor suppressor and indicates that TAp73 exerts this function, at least partially, by regulation of cellular metabolism.
Collapse
Affiliation(s)
- Massimiliano Agostini
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | | | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandro Rufini
- Department of Cancer Studies, CRUK Leicester Cancer Centre, University of Leicester, Leicester, UK
| |
Collapse
|
123
|
Rodríguez N, Peláez A, Barderas R, Domínguez G. Clinical implications of the deregulated TP73 isoforms expression in cancer. Clin Transl Oncol 2017; 20:827-836. [PMID: 29230693 DOI: 10.1007/s12094-017-1802-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/09/2017] [Indexed: 01/24/2023]
Abstract
TP73 is a member of the TP53 family whose expression has been observed altered in most human cancers and associated with the prognosis. TP73 translates into a complex number of isoforms with both oncogenic and tumour-suppressor functions and presents a complex cross-talk with other members of the family (TP53 and TP63). In this revision, we focus on the evidence that may support TP73 variants as prognostic markers in cancer. Nowadays, most publications in this topic highlight the association between overexpression of the oncogenic variants and failure to respond to chemotherapy and/or shorter survival. In addition, we comment on the putative possibilities that the detection through a liquid biopsy of TP73 variants may provide, and finally, the significance of determining the value of the combined alteration of the TP53 family members in the clinical setting.
Collapse
Affiliation(s)
- N Rodríguez
- Servicio de Oncología Médica, Hospital Universitario La Paz, CIBERONC, Madrid, Spain
| | - A Peláez
- Servicio de Anatomía Patológica and Molecular Pathology and Therapeutic Targets Group, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - R Barderas
- UFIEC, ISCIII, Majadahonda, Madrid, Spain
| | - G Domínguez
- Departamento de Medicina, Facultad de Medicina, Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-Universidad Autónoma de Madrid, CIBERONC, Madrid, Spain.
| |
Collapse
|
124
|
Rahmani M, Talebi M, Hagh MF, Feizi AAH, Solali S. Aberrant DNA methylation of key genes and Acute Lymphoblastic Leukemia. Biomed Pharmacother 2017; 97:1493-1500. [PMID: 29793312 DOI: 10.1016/j.biopha.2017.11.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/27/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023] Open
Abstract
DNA methylation is a dynamic process influencing gene expression by altering either coding or non-coding loci. Despite advances in treatment of Acute Lymphoblastic Leukemia (ALL); relapse occurs in approximately 20% of patients. Nowadays, epigenetic factors are considered as one of the most effective mechanisms in pathogenesis of malignancies. These factors are reversible elements which can be potentially regarded as therapy targets and disease prognosis. DNA methylation, which primarily serves as transcriptional suppressor, mostly occurs in CpG islands of the gene promoter regions. This was shown as a key epigenetic factor in inactivating various tumor suppressor genes during cancer initiation and progression. We aimed to review methylation status of key genes involved in hematopoietic malignancies such as IKZF1, CDKN2B, TET2, CYP1B1, SALL4, DLC1, DLX family, TP73, PTPN6, and CDKN1C; and their significance in pathogenesis of ALL. The DNA methylation alterations in promoter regions of the genes have been shown to play crucial roles in tumorigenesis. Methylation -based inactivation of these genes has also been reported as associated with prognosis in acute leukemia. In this review, we also addressed the association of gene expression and methylation pattern in ALL patients.
Collapse
Affiliation(s)
- Mina Rahmani
- Department of Immunology, Division of Hematology and Transfusion Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Farshdousti Hagh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saeed Solali
- Department of Immunology, Division of Hematology and Transfusion Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
125
|
Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ 2017; 25:104-113. [PMID: 29149101 DOI: 10.1038/cdd.2017.169] [Citation(s) in RCA: 817] [Impact Index Per Article: 116.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 01/10/2023] Open
Abstract
The tumour suppressor gene TP53 is mutated in ~50% of human cancers. In addition to its function in tumour suppression, p53 also plays a major role in the response of malignant as well as nontransformed cells to many anticancer therapeutics, particularly those that cause DNA damage. P53 forms a homotetrameric transcription factor that is reported to directly regulate ~500 target genes, thereby controlling a broad range of cellular processes, including cell cycle arrest, cell senescence, DNA repair, metabolic adaptation and cell death. For a long time, induction of apoptotic death in nascent neoplastic cells was regarded as the principal mechanism by which p53 prevents tumour development. This concept has, however, recently been challenged by the findings that in striking contrast to Trp53-deficient mice, gene-targeted mice that lack the critical effectors of p53-induced apoptosis do not develop tumours spontaneously. Remarkably, even mice lacking all mediators critical for p53-induced apoptosis, G1/S boundary cell cycle arrest and cell senescence do not develop any tumours spontaneously. In this review we discuss current understanding of the mechanisms by which p53 induces cell death and how this affects p53-mediated tumour suppression and the response of malignant cells to anticancer therapy.
Collapse
Affiliation(s)
- Brandon J Aubrey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ana Janic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
126
|
Nemajerova A, Amelio I, Gebel J, Dötsch V, Melino G, Moll UM. Non-oncogenic roles of TAp73: from multiciliogenesis to metabolism. Cell Death Differ 2017; 25:144-153. [PMID: 29077094 PMCID: PMC5729534 DOI: 10.1038/cdd.2017.178] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 01/24/2023] Open
Abstract
The p53 family of transcription factors (p53, p63 and p73) covers a wide range of functions critical for development, homeostasis and health of mammals across their lifespan. Beside the well-established tumor suppressor role, recent evidence has highlighted novel non-oncogenic functions exerted by p73. In particular, p73 is required for multiciliated cell (MCC) differentiation; MCCs have critical roles in brain and airways to move fluids across epithelial surfaces and to transport germ cells in the reproductive tract. This novel function of p73 provides a unifying cellular mechanism for the disparate inflammatory and immunological phenotypes of p73-deficient mice. Indeed, mice with Trp73 deficiency suffer from hydrocephalus, sterility and chronic respiratory tract infections due to profound defects in ciliogenesis and complete loss of mucociliary clearance since MCCs are essential for cleaning airways from inhaled pollutants, pathogens and allergens. Cross-species genomic analyses and functional rescue experiments identify TAp73 as the master transcriptional integrator of ciliogenesis, upstream of previously known central nodes. In addition, TAp73 shows a significant ability to regulate cellular metabolism and energy production through direct transcriptional regulation of several metabolic enzymes, such as glutaminase-2 and glucose-6 phosphate dehydrogenase. This recently uncovered role of TAp73 in the regulation of cellular metabolism strongly affects oxidative balance, thus potentially influencing all the biological aspects associated with p73 function, including development, homeostasis and cancer. Although through different mechanisms, p63 isoforms also contribute to regulation of cellular metabolism, thus indicating a common route used by all family members to control cell fate. At the structural level, the complexity of p73's function is further enhanced by its ability to form heterotetramers with some p63 isoforms, thus indicating the existence of an intrafamily crosstalk that determines the global outcome of p53 family function. In this review, we have tried to summarize all the recent evidence that have emerged on the novel non-oncogenic roles of p73, in an attempt to provide a unified view of the complex function of this gene within its family.
Collapse
Affiliation(s)
- Alice Nemajerova
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ivano Amelio
- Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, PO Box 138, Leicester LE1 9HN, UK
| | - Jakob Gebel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, PO Box 138, Leicester LE1 9HN, UK.,Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', Rome 00133, Italy
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
127
|
Monti P, Ghiorzo P, Menichini P, Foggetti G, Queirolo P, Izzotti A, Fronza G. TP63 mutations are frequent in cutaneous melanoma, support UV etiology, but their role in melanomagenesis is unclear. Oncol Rep 2017; 38:1985-1994. [PMID: 28849221 PMCID: PMC5652947 DOI: 10.3892/or.2017.5903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
In contrast to TP53, cancer development is rarely associated with mutations in the TP63 and TP73 genes. Recently, next generation sequencing analysis revealed that TP63 mutations are frequent, specifically in cutaneous melanomas. Cutaneous melanoma represents 4% of skin cancers but it is responsible for 80% of skin cancer related deaths. In the present study, we first determined whether all three members of the P53 family of transcription factors were found mutated in cutaneous melanomas by retrieving all TP53, TP63 and TP73 mutations from cBioPortal (http://www.cbioportal.org/). TP53 and TP63 were frequently mutated [15.0% (91/605) and 14.7% (89/605), respectively], while TP73 [1.5% (9/605)] was more rarely mutated (p<0.0001). A UV-mutation fingerprint was recognized for TP63 and TP73 genes. Then, we tried to evaluate the potential role of TP63 mutations as drivers or passengers in the tumorigenic process. In the former case, the amino acid substitutions should cause significant functional consequences on the main biochemical activity of the P63 protein, namely transactivation. The predicted effects of specific amino acid substitutions by two bioinformatics tools were rather different. Using a yeast-based functional assay, the observed hotspot mutant R379CP63 protein exhibited a substantial residual activity compared to the wild-type (>70%). This result does not support a major role of the mutant P63 protein in melanomagenesis while it is still consistent with the TP63 gene being a recorder of UV exposure. The TP63 mutation spectrum from cutaneous melanomas, when compared with that observed at the germinal level in patients affected by P63-associated diseases [ectodermal dysplasia syndromes, (EDs)], revealed significant differences. The TP63 mutations were more frequent at CpGs sites (p<0.0001) in EDs and at PyPy sites (p<0.0001) in cutaneous melanomas. The two spectra differed significantly (p<0.0001). We conclude that TP63 mutations are frequent in cutaneous melanoma, support UV etiology, but their role in melanomagenesis is unclear.
Collapse
Affiliation(s)
- Paola Monti
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genova, I-16132 Genova, Italy
- Genetics of Rare Cancers Unit, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Paola Menichini
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Giorgia Foggetti
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Paola Queirolo
- Medical Oncology Unit, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Alberto Izzotti
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
- Department of Health Sciences, University of Genova, I-16132 Genova, Italy
| | - Gilberto Fronza
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| |
Collapse
|
128
|
Gebel J, Tuppi M, Krauskopf K, Coutandin D, Pitzius S, Kehrloesser S, Osterburg C, Dötsch V. Control mechanisms in germ cells mediated by p53 family proteins. J Cell Sci 2017:jcs.204859. [PMID: 28794013 DOI: 10.1242/jcs.204859] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Germ cells are totipotent and, in principle, immortal as they are the source for new germ cells in each generation. This very special role requires tight quality control systems. The p53 protein family constitutes one of the most important quality surveillance systems in cells. Whereas p53 has become famous for its role as the guardian of the genome in its function as the most important somatic tumor suppressor, p63 has been nicknamed 'guardian of the female germ line'. p63 is strongly expressed in resting oocytes and responsible for eliminating those that carry DNA double-strand breaks. The third family member, p73, acts later during oocyte and embryo development by ensuring correct assembly of the spindle assembly checkpoint. In addition to its role in the female germ line, p73 regulates cell-cell contacts between developing sperm cells and supporting somatic cells in the male germ line. Here, we review the involvement of the p53 protein family in the development of germ cells with a focus on quality control in the female germ line and discuss medical implications for cancer patients.
Collapse
Affiliation(s)
- Jakob Gebel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| | - Marcel Tuppi
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| | - Katharina Krauskopf
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| | - Daniel Coutandin
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| | - Susanne Pitzius
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| | - Sebastian Kehrloesser
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| | - Christian Osterburg
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Max von Laue-Str. 9, Frankfurt am Main 60438, Germany
| |
Collapse
|
129
|
Abstract
Since its discovery in 1979, p53 has been on the forefront of cancer research. It is considered a master gene of cancer suppression and is found mutated in around 50% of all human tumors. In addition, the progressive identification of p53-related transcription factors p63 and p73 as well as their multiple isoforms have added further layers of complexity to an already dense network. Among the numerous models used to unravel the p53 family mysteries, S. cerevisiae has been particularly useful. This seemingly naive model allows the expression of a functional human p53 and thus the assessment of p53 intrinsic transcriptional activity. The aim of this article is to review the various contributions that the budding yeast has made to the understanding of p53, p63 and p73 biology and to envision new possible directions for yeast-based assays in the field of cancer as well as other p53-family-related diseases.
Collapse
|
130
|
Li W, Wang SS, Deng J, Tang JX. Association of p73 gene G4C14-A4T14 polymorphism and MDM2 gene SNP309 with non-small cell lung cancer risk in a Chinese population. Oncol Lett 2017; 14:1817-1822. [PMID: 28789416 DOI: 10.3892/ol.2017.6327] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/17/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the association of p73 G4C14-A4T14 polymorphism and murine double minute 2 (MDM2) 309 T/G single nucleotide polymorphisms (SNPs) with the risk of developing non-small cell lung cancer (NSCLC) in Sothern China. The p73 and MDM2 genotypes of peripheral blood DNA from 186 patients with NSCLC and 196 normal controls were detected by polymerase chain reaction (PCR) with confronting two-pair primers (CTPP) and high resolution melting (HRM), respectively. The results of genotyping were consistent with those of direct sequencing. The p73 AT/AT [odds ratio (OR)=0.46; 95% confidence interval (CI)=0.22-0.97] and MDM2 TT (OR=0.48; 95% CI=0.26-0.86) genotypes were associated with a decreased risk of developing NSCLC compared with that of the p73 GC/GC and MDM2 GG genotypes, respectively. In addition, the interaction between the p73 and MDM2 polymorphisms reduced the risk of developing NSCLC in multiple ways (OR=0.13; 95% CI=0.03-0.59) for subjects carrying both the p73 AT/AT and MDM2 TT genotypes. Therefore, the SNP in p73 G4C14-A4T14 and the MDM2 309 polymorphism may be markers of genetic susceptibility to NSCLC in a Chinese population, and there is a possible gene-gene interaction involved in the incidence of NSCLC.
Collapse
Affiliation(s)
- Wen Li
- Key Laboratory of Green Packaging and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, Hunan 412007, P.R. China
| | - Shuang Shuang Wang
- Key Laboratory of Green Packaging and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, Hunan 412007, P.R. China
| | - Jing Deng
- College of Packaging and Material Engineering, Hunan University of Technology, Zhuzhou, Hunan 412007, P.R. China
| | - Jian Xin Tang
- Key Laboratory of Green Packaging and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, Hunan 412007, P.R. China
| |
Collapse
|
131
|
Rao AKDM, Manikandan M, Arunkumar G, Revathidevi S, Vinothkumar V, Arun K, Tiwary BK, Rajkumar KS, Rajaraman R, Munirajan AK. Prevalence of p53 codon 72, p73 G4C14-A4T14 and MDM2 T309G polymorphisms and its association with the risk of oral cancer in South Indians. GENE REPORTS 2017. [DOI: 10.1016/j.genrep.2017.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
132
|
Mori N, Ohwashi-Miyazaki M, Yoshinaga K, Okada M, Shiseki M, Motoji T, Tanaka J. Tumor suppressor gene methylation on the short arm of chromosome 1 in chronic myelogenous leukemia. Eur J Haematol 2017; 98:467-477. [PMID: 28129457 DOI: 10.1111/ejh.12857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVES We previously reported loss of heterozygosity on 1p in chronic myelogenous leukemia (CML). We analyzed promoter methylation and mutation of tumor suppressor genes on 1p36 in CML. METHODS We performed methylation-specific PCR (MS-PCR) analysis of the PRDM2, RUNX3, and TP73 genes in 61 patients with CML (43 chronic phase, CP; two accelerated phase; and 16 blast crisis, BC). Oxidative MS-PCR, PCR-single-strand conformation polymorphism, and real-time reverse transcriptase PCR were also analyzed. K-562 cells were grown in the presence of 5-Aza-dC and trichostatin A. RESULTS Methylation of the PRDM2, RUNX3, and TP73 genes was detected in 24/60 (40%), 21/61 (34%), and 28/60 (47%) patients, respectively. Methylation of all three genes was detected in 19/59 (32%) patients. Methylation was more frequent in BC than in CP. Oxidative MS-PCR analysis detected 5-mC in the PRDM2, RUNX3, and TP73 genes in 10/22 (45%), 15/21 (71%), and 16/26 (62%) samples with methylation detected by MS-PCR, respectively. Decreased expression was observed in several samples with methylation, while no mutations were found in the genes. Treatment of K-562 cells induced growth suppression, demethylation, and reexpression of the PRDM2 and RUNX3 genes. CONCLUSION Multiple tumor suppressor genes on 1p were inactivated in CML by methylation.
Collapse
Affiliation(s)
- Naoki Mori
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | | | - Kentaro Yoshinaga
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Michiko Okada
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masayuki Shiseki
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshiko Motoji
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Junji Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
133
|
Abstract
The role of p53 as “a guardian of the genome” has been well established in somatic cells. However, its role in pluripotent stem cells remains much more elusive. Here, we discuss research progress in understanding the role of p53 in pluripotent stem cells and in pluripotent stem cell-like cancer stem cells. The p53 protein, which plays a key role in embryonic stem cells, was first discovered in 2005. Landmark studies of p53-related reprogramming elucidated this protein’s importance in induced pluripotent stem cells in 2009. The p53-related safety concerns in pluripotent stem cells have been raised in stem cell-based therapy although the use of iPSCs in therapeutic application is promising. Because cancer stem cells have profiles similar to those of pluripotent stem cells, we also describe potential strategies for studies in cancer stem cells and cancer treatments. The new discoveries of p53 family proteins in pluripotent stem cells have made possible stable progress in stem cell transplantation efficiency and safety, as well as treatment strategies targeting cancer stem cells based on pluripotent stem cell technology.
Collapse
Affiliation(s)
- Tongxiang Lin
- Stem Cell Research Center, College of Bee Science, Fujian Agriculture and Forestry University, 15 ShangXiaDian Rd, Fuzhou, Fujian, 350002, China. .,Center for Regenerative and Translational Medicine, The Second Affiliated Hospital (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou University of Chinese Medicine, 111 Dade Rd, Guangzhou, Guangdong, 510120, China.
| | - Yi Lin
- Stem Cell Research Center, College of Bee Science, Fujian Agriculture and Forestry University, 15 ShangXiaDian Rd, Fuzhou, Fujian, 350002, China.
| |
Collapse
|
134
|
Ge L, Yang Y, Sun Y, Xu W, Lu D, Su B. P73 G4C14-to-A4T14 polymorphism is associated with survival in advanced non-small cell lung cancer patients. Thorac Cancer 2017; 8:63-72. [PMID: 28134496 PMCID: PMC5334305 DOI: 10.1111/1759-7714.12397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/04/2016] [Accepted: 09/11/2016] [Indexed: 12/01/2022] Open
Abstract
Background p73, a structural and functional homolog of p53, plays an important role in modulating cell cycle arrest. This study investigated the association between p73 G4C14‐to‐A4T14 polymorphism and survival outcomes in a Chinese population of advanced non‐small cell lung cancer (NSCLC) patients treated with platinum agents. Methods The p73 G4C14‐to‐A4T14 polymorphism was genotyped using DNA from blood samples of advanced NSCLC patients (642 in the discovery set and 330 in the replication set). The relationship of the p73 G4C14‐to‐A4T14 polymorphism with clinical outcomes was analyzed. Results Compared with the GC/GC genotype, the genotypes containing AT allele (GC/AT + AT/AT genotypes) were associated with significantly prolonged overall survival (P = 0.040) in the discovery set and after pooling results from the replication set. Stratification analysis revealed that the association was more pronounced in subjects who were older (P = 0.001), male (P = 0.007), smokers (P = 0.006), had a low Eastern Cooperative Oncology Group performance status (P = 0.001), in tumor node metastasis stage IV (P = 0.008), and with adenocarcinoma (P = 0.002). The objective response rates of patients with GC/AT + AT/AT genotypes were statistically higher than those with the GC/GC genotype (P = 0.047). Conclusion Our findings suggest that the p73 G4C14‐to‐A4T14 polymorphism may be related to survival outcome in advanced NSCLC patients.
Collapse
Affiliation(s)
- Lei Ge
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yifeng Sun
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Xu
- Department of Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Bo Su
- Department of Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
135
|
Wang W, Bao T, Zeng X, Xiong H, Wen W, Zhang X, Wang S. Ultrasensitive electrochemical DNA biosensor based on functionalized gold clusters/graphene nanohybrids coupling with exonuclease III-aided cascade target recycling. Biosens Bioelectron 2016; 91:183-189. [PMID: 28006687 DOI: 10.1016/j.bios.2016.12.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 02/08/2023]
Abstract
In this work, a novel and ultrasensitive electrochemical biosensor was constructed for DNA detection based on functionalized gold clusters/graphene nanohybrids (AuNCs/GR nanobybrids) and exonuclease III (Exo III)-aided cascade target recycling. By utilizing the capacity of GR as universal template, different metal nanoclusters including AuNCs/GR nanobybrids and PtNCs/GR nanohybrids were synthesized through convenient ultrasonic method. Exo III-aided cascade recycling was initiated by target DNA, generating the final cleavage product (S2), which acted as a linkage between capture probe and the functionalized metal nanoclusters/GR conjugates in the construction of the biosensor. The AuNCs/GR-DNA-enzyme conjugates acted as interfaces of enzyme-catalyzed silver deposition reaction, achieving DNA detection ranging from 0.02 fM to 20 pM with a detection limit of 0.057 fM. In addition, PtNCs/GR-DNA conjugates presented peroxidase-like activity and the functionalized PtNCs/GR nanohybrids-based electrochemical biosensor also realized DNA detection by catalyzing the 3,3',5,5'-tetramethylbenzidine-hydrogen peroxide (TMB-H2O2) system to produce electrochemical signal. This metal clusters/GR-based multiple-amplified electrochemical biosensor provided an universal method for DNA detection.
Collapse
Affiliation(s)
- Wei Wang
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Ting Bao
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Xi Zeng
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Huayu Xiong
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Wei Wen
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Xiuhua Zhang
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China
| | - Shengfu Wang
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules & College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, PR China.
| |
Collapse
|
136
|
Armstrong SR, Wu H, Wang B, Abuetabh Y, Sergi C, Leng RP. The Regulation of Tumor Suppressor p63 by the Ubiquitin-Proteasome System. Int J Mol Sci 2016; 17:ijms17122041. [PMID: 27929429 PMCID: PMC5187841 DOI: 10.3390/ijms17122041] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/23/2016] [Accepted: 11/30/2016] [Indexed: 12/18/2022] Open
Abstract
The protein p63 has been identified as a homolog of the tumor suppressor protein p53 and is capable of inducing apoptosis, cell cycle arrest, or senescence. p63 has at least six isoforms, which can be divided into two major groups: the TAp63 variants that contain the N-terminal transactivation domain and the ΔNp63 variants that lack the N-terminal transactivation domain. The TAp63 variants are generally considered to be tumor suppressors involved in activating apoptosis and suppressing metastasis. ΔNp63 variants cannot induce apoptosis but can act as dominant negative inhibitors to block the function of TAp53, TAp73, and TAp63. p63 is rarely mutated in human tumors and is predominately regulated at the post-translational level by phosphorylation and ubiquitination. This review focuses primarily on regulation of p63 by the ubiquitin E-3 ligase family of enzymes via ubiquitination and proteasome-mediated degradation, and introduces a new key regulator of the p63 protein.
Collapse
Affiliation(s)
- Stephen R Armstrong
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| | - Hong Wu
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| | - Benfan Wang
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| | - Yasser Abuetabh
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada.
| | - Roger P Leng
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| |
Collapse
|
137
|
Kehrloesser S, Osterburg C, Tuppi M, Schäfer B, Vousden KH, Dötsch V. Intrinsic aggregation propensity of the p63 and p73 TI domains correlates with p53R175H interaction and suggests further significance of aggregation events in the p53 family. Cell Death Differ 2016; 23:1952-1960. [PMID: 27447112 PMCID: PMC5136486 DOI: 10.1038/cdd.2016.75] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/19/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022] Open
Abstract
The high percentage of p53 missense mutations found in cancer has been attributed to mutant acquired oncogenic gain of functions. Different aspects of these tumour-promoting functions are caused by repression of the transcriptional activity of p53 family members p63 and p73. A subset of frequently occurring p53 mutations results in thermodynamic destabilisation of the DNA-binding domain (DBD) rendering this domain highly unstable. These conformational mutants (such as p53R175H) have been suggested to directly bind to p63 and p73 via a co-aggregation mechanism mediated by their DBDs. Although the DBDs of p63 and p73 are in fact not sufficient for the interaction as shown previously, we demonstrate here that the transactivation inhibitory (TI) domains within the α-isoform-specific C termini of p63 and p73 are essential for binding to p53R175H. Hence, the closed dimeric conformation of inactive TAp63α that renders the TI domain inaccessible prevents efficient interaction. We further show that binding to p53R175H correlates with an intrinsic aggregation propensity of the tetrameric α-isoforms conferred by an openly accessible TI domain again supporting interaction via a co-aggregation mechanism.
Collapse
Affiliation(s)
- Sebastian Kehrloesser
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Christian Osterburg
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Marcel Tuppi
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Birgit Schäfer
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University Frankfurt, Frankfurt/Main, Germany
| | | | - Volker Dötsch
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University Frankfurt, Frankfurt/Main, Germany
| |
Collapse
|
138
|
Abstract
Since its discovery more than three decades ago, tumor suppressor p53 has been shown to play pivotal roles in both maintaining genomic integrity and tumor suppression. p53 functions as a transcription factor responding to a multitude of cellular stressors, regulating the transcription of many genes involved in cell-cycle arrest, senescence, autophagy, and apoptosis. Extensive work has revealed that p53 is one of the most commonly mutated tumor suppressor genes. The last three decades have demonstrated that p53 activity is controlled through transcriptional regulation and posttranslational modifications. However, evolving work is now uncovering that p53, and other p53 family members, are post-transcriptionally regulated by multiple RNA-binding proteins (RBPs). Understanding the regulation of p53 by RBPs may potentially open up the possibility for cancer therapeutic intervention. This review focuses on the posttranscriptional regulation of p53, and p53 family members, by RNA binding proteins and the reciprocal feedback pathways between several RNA-biding proteins modulating p53, and p53 family members.
Collapse
Affiliation(s)
- Chris Lucchesi
- Comparative Oncology Laboratory, School of Veterinary Medicine, School of Medicine, University of California at Davis, Davis, California 95616, USA
| | - Jin Zhang
- Comparative Oncology Laboratory, School of Veterinary Medicine, School of Medicine, University of California at Davis, Davis, California 95616, USA
| | - Xinbin Chen
- Comparative Oncology Laboratory, School of Veterinary Medicine, School of Medicine, University of California at Davis, Davis, California 95616, USA
| |
Collapse
|
139
|
Olivos DJ, Mayo LD. Emerging Non-Canonical Functions and Regulation by p53: p53 and Stemness. Int J Mol Sci 2016; 17:ijms17121982. [PMID: 27898034 PMCID: PMC5187782 DOI: 10.3390/ijms17121982] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/15/2023] Open
Abstract
Since its discovery nearly 40 years ago, p53 has ascended to the forefront of investigated genes and proteins across diverse research disciplines and is recognized most exclusively for its role in cancer as a tumor suppressor. Levine and Oren (2009) reviewed the evolution of p53 detailing the significant discoveries of each decade since its first report in 1979. In this review, we will highlight the emerging non-canonical functions and regulation of p53 in stem cells. We will focus on general themes shared among p53's functions in non-malignant stem cells and cancer stem-like cells (CSCs) and the influence of p53 on the microenvironment and CSC niche. We will also examine p53 gain of function (GOF) roles in stemness. Mutant p53 (mutp53) GOFs that lead to survival, drug resistance and colonization are reviewed in the context of the acquisition of advantageous transformation processes, such as differentiation and dedifferentiation, epithelial-to-mesenchymal transition (EMT) and stem cell senescence and quiescence. Finally, we will conclude with therapeutic strategies that restore wild-type p53 (wtp53) function in cancer and CSCs, including RING finger E3 ligases and CSC maintenance. The mechanisms by which wtp53 and mutp53 influence stemness in non-malignant stem cells and CSCs or tumor-initiating cells (TICs) are poorly understood thus far. Further elucidation of p53's effects on stemness could lead to novel therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- David J Olivos
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Lindsey D Mayo
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
140
|
Alhosin M, Omran Z, Zamzami MA, Al-Malki AL, Choudhry H, Mousli M, Bronner C. Signalling pathways in UHRF1-dependent regulation of tumor suppressor genes in cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:174. [PMID: 27839516 PMCID: PMC5108085 DOI: 10.1186/s13046-016-0453-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/02/2016] [Indexed: 12/13/2022]
Abstract
Epigenetic silencing of tumor suppressor genes (TSGs) through DNA methylation and histone changes is a main hallmark of cancer. Ubiquitin-like with PHD and RING Finger domains 1 (UHRF1) is a potent oncogene overexpressed in various solid and haematological tumors and its high expression levels are associated with decreased expression of several TSGs including p16INK4A, BRCA1, PPARG and KiSS1. Using its several functional domains, UHRF1 creates a strong coordinated dialogue between DNA methylation and histone post-translation modification changes causing the epigenetic silencing of TSGs which allows cancer cells to escape apoptosis. To ensure the silencing of TSGs during cell division, UHRF1 recruits several enzymes including histone deacetylase 1 (HDAC1), DNA methyltransferase 1 (DNMT1) and histone lysine methyltransferases G9a and Suv39H1 to the right place at the right moment. Several in vitro and in vivo works have reported the direct implication of the epigenetic player UHRF1 in tumorigenesis through the repression of TSGs expression and suggested UHRF1 as a promising target for cancer treatment. This review describes the molecular mechanisms underlying UHRF1 regulation in cancer and discusses its importance as a therapeutic target to induce the reactivation of TSGs and subsequent apoptosis.
Collapse
Affiliation(s)
- Mahmoud Alhosin
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia. .,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia. .,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia. .,Biochemistry Department, Faculty of Sciences, Cancer and Mutagenesis Unit, King Fahd Centre for Medical Research, King Abdulaziz University, P. O. Box 80203, Jeddah, 21589, Saudi Arabia.
| | - Ziad Omran
- College of Pharmacy, Umm Al-Qura University, 21955, Makkah, Kingdom of Saudi Arabia
| | - Mazin A Zamzami
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman L Al-Malki
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Marc Mousli
- Laboratoire de Biophotonique et Pharmacologie, UMR 7213 CNRS, Université de Strasbourg, Faculté de pharmacie, 74 route du Rhin, 67401, Illkirch, France
| | - Christian Bronner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964 CNRS UMR 7104, Université de Strasbourg, 1 rue Laurent Fries, 67404, Illkirch, France.
| |
Collapse
|
141
|
Mechanism of TAp73 inhibition by ΔNp63 and structural basis of p63/p73 hetero-tetramerization. Cell Death Differ 2016; 23:1930-1940. [PMID: 27716744 DOI: 10.1038/cdd.2016.83] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/28/2016] [Accepted: 07/12/2016] [Indexed: 11/09/2022] Open
Abstract
Members of the p53 tumor-suppressor family are expressed as multiple isoforms. Isoforms with an N-terminal transactivation domain are transcriptionally active, while those ones lacking this domain often inhibit the transcriptional activity of other family members. In squamous cell carcinomas, the high expression level of ΔNp63α inhibits the tumor-suppressor function of TAp73β. This can in principle be due to blocking of the promoter or by direct interaction between both proteins. p63 and p73 can hetero-oligomerize through their tetramerization domains and a hetero-tetramer consisting of two p63 and two p73 molecules is thermodynamically more stable than both homo-tetramers. Here we show that cells expressing both p63 and p73 exist in mouse epidermis and hair follicle and that hetero-tetramer complexes can be detected by immunoprecipitation in differentiating keratinocytes. Through structure determination of the hetero-tetramer, we reveal why this hetero-tetramer is the thermodynamically preferred species. We have created mutants that exclusively form either hetero-tetramers or homo-tetramers, allowing to investigate the function of these p63/p73 hetero-tetramers. Using these tools, we show that inhibition of TAp73β in squamous cell carcinomas is due to promoter squelching and not direct interaction.
Collapse
|
142
|
Costa DCF, de Oliveira GAP, Cino EA, Soares IN, Rangel LP, Silva JL. Aggregation and Prion-Like Properties of Misfolded Tumor Suppressors: Is Cancer a Prion Disease? Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a023614. [PMID: 27549118 DOI: 10.1101/cshperspect.a023614] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Prion diseases are disorders that share several characteristics that are typical of many neurodegenerative diseases. Recently, several studies have extended the prion concept to pathological aggregation in malignant tumors involving misfolded p53, a tumor-suppressor protein. The aggregation of p53 and its coaggregation with p53 family members, p63 and p73, have been shown. Certain p53 mutants exert a dominant-negative regulatory effect on wild-type (WT) p53. The basis for this dominant-negative effect is that amyloid-like mutant p53 converts WT p53 into an aggregated species, leading to a gain-of-function (GoF) phenotype and the loss of its tumor-suppressor function. Recently, it was shown that p53 aggregates can be internalized by cells and can coaggregate with endogenous p53, corroborating the prion-like properties of p53 aggregates. The prion-like behavior of oncogenic p53 mutants provides an explanation for its dominant-negative and GoF properties, including the high metastatic potential of cancer cells carrying p53 mutations. The inhibition of p53 aggregation appears to represent a promising target for therapeutic intervention in patients with malignant tumors.
Collapse
Affiliation(s)
- Danielly C F Costa
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil Instituto Nacional de Ciência e Tecnologia (INCT) de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil Instituto de Nutrição, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20550-013, Brazil
| | - Guilherme A P de Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil Instituto Nacional de Ciência e Tecnologia (INCT) de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Elio A Cino
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil Instituto Nacional de Ciência e Tecnologia (INCT) de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Iaci N Soares
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil Instituto Nacional de Ciência e Tecnologia (INCT) de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Luciana P Rangel
- Instituto Nacional de Ciência e Tecnologia (INCT) de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Jerson L Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil Instituto Nacional de Ciência e Tecnologia (INCT) de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| |
Collapse
|
143
|
He Z, Agostini M, Liu H, Melino G, Simon HU. p73 regulates basal and starvation-induced liver metabolism in vivo. Oncotarget 2016; 6:33178-90. [PMID: 26375672 PMCID: PMC4741757 DOI: 10.18632/oncotarget.5090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/26/2015] [Indexed: 12/30/2022] Open
Abstract
As a member of the p53 gene family, p73 regulates cell cycle arrest, apoptosis, neurogenesis, immunity and inflammation. Recently, p73 has been shown to transcriptionally regulate selective metabolic enzymes, such as cytochrome c oxidase subunit IV isoform 1, glucose 6-phosphate dehydrogenase and glutaminase-2, resulting in significant effects on metabolism, including hepatocellular lipid metabolism, glutathione homeostasis and the pentose phosphate pathway. In order to further investigate the metabolic effect of p73, here, we compared the global metabolic profile of livers from p73 knockout and wild-type mice under both control and starvation conditions. Our results show that the depletion of all p73 isoforms cause altered lysine metabolism and glycolysis, distinct patterns for glutathione synthesis and Krebs cycle, as well as an elevated pentose phosphate pathway and abnormal lipid accumulation. These results indicate that p73 regulates basal and starvation-induced fuel metabolism in the liver, a finding that is likely to be highly relevant for metabolism-associated disorders, such as diabetes and cancer.
Collapse
Affiliation(s)
- Zhaoyue He
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Massimiliano Agostini
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - He Liu
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
144
|
Baudot AD, Crighton D, O'Prey J, Somers J, Sierra Gonzalez P, Ryan KM. p53 directly regulates the glycosidase FUCA1 to promote chemotherapy-induced cell death. Cell Cycle 2016; 15:2299-308. [PMID: 27315169 PMCID: PMC5004703 DOI: 10.1080/15384101.2016.1191714] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/11/2016] [Accepted: 05/14/2016] [Indexed: 12/16/2022] Open
Abstract
p53 is a central factor in tumor suppression as exemplified by its frequent loss in human cancer. p53 exerts its tumor suppressive effects in multiple ways, but the ability to invoke the eradication of damaged cells by programmed cell death is considered a key factor. The ways in which p53 promotes cell death can involve direct activation or engagement of the cell death machinery, or can be via indirect mechanisms, for example though regulation of ER stress and autophagy. We present here another level of control in p53-mediated tumor suppression by showing that p53 activates the glycosidase, FUCA1, a modulator of N-linked glycosylation. We show that p53 transcriptionally activates FUCA1 and that p53 modulates fucosidase activity via FUCA1 up-regulation. Importantly, we also report that chemotherapeutic drugs induce FUCA1 and fucosidase activity in a p53-dependent manner. In this context, while we found that over-expression of FUCA1 does not induce cell death, RNAi-mediated knockdown of endogenous FUCA1 significantly attenuates p53-dependent, chemotherapy-induced apoptotic death. In summary, these findings add an additional component to p53s tumor suppressive response and highlight another mechanism by which the tumor suppressor controls programmed cell death that could potentially be exploited for cancer therapy.
Collapse
Affiliation(s)
- Alice D. Baudot
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | - Diane Crighton
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | - Jim O'Prey
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | - Joanna Somers
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | | | - Kevin M. Ryan
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| |
Collapse
|
145
|
Xie T, Yu CH, Zheng Y, A ZC. The polymorphism G4C14-to-A4T14 in p73 gene may affect the susceptibility to male infertility with severe spermatogenesis impairment in Chinese population. Eur J Obstet Gynecol Reprod Biol 2016; 204:74-7. [PMID: 27525684 DOI: 10.1016/j.ejogrb.2016.07.508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/07/2016] [Accepted: 07/26/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The aim of this study was to explore the association between the polymorphism G4C14-to-A4T14 in the p73 gene and male infertility with severe spermatogenesis impairment in Chinese population. STUDY DESIGN Three hundreds and one infertile patients with severe spermatogenesis impairment (including azoospermia and severe oligospermia) and 252 fertile men were recruited in this study. The polymorphism G4C14-to-A4T14 in the p73 gene was genotyped using polymerase chain reaction and restriction fragment length polymorphism assay. The differences in allelic and genotypic frequencies between patients and controls were evaluated by chi-square test. RESULTS The frequency of allele AT (28.9% vs. 22.4%, P=0.017, OR=1.41, 95% CI=1.07-1.85) in patients with severe spermatogenesis impairment was significantly higher than that in controls, whereas the genotype GC/GC was significantly decreased in patients compared with controls (48.5% vs. 59.1%, P=0.048, OR=0.65, 95% CI=0.46-0.91). CONCULUSION The findings of this study suggested that the polymorphism G4C14-to-A4T14 in p73 gene might be associated with severe spermatogenesis impairment and could affect the susceptibility to male infertility with severe spermatogenesis impairment in Chinese population.
Collapse
Affiliation(s)
- Ting Xie
- College of Basic Medicine, Dali University, Dali, 671000, China
| | - Cheng-He Yu
- College of Basic Medicine, Dali University, Dali, 671000, China; Department of Reproductive Medicine, Affiliated Hospital of Dali University, Dali, 671000, China
| | - Ye Zheng
- Department of Genetics, College of Agriculture and Biology, Dali University, Dali, 671003, China
| | - Zhou-Cun A
- College of Basic Medicine, Dali University, Dali, 671000, China; Department of Genetics, College of Agriculture and Biology, Dali University, Dali, 671003, China.
| |
Collapse
|
146
|
P63 expression as a biomarker discriminating giant cell tumor of bone from other giant cell-rich bone lesions. Pathol Res Pract 2016; 212:876-879. [PMID: 27473669 DOI: 10.1016/j.prp.2016.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/12/2016] [Accepted: 07/12/2016] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Giant cell tumor of bone (GCTOB) is a locally aggressive neoplasm that accounts for 5% of all primary bone tumors. This tumor overlaps in histopathologic and radiographic presentations with different malignant, benign, and metabolic giant cell-rich lesions. The purpose of this study is to evaluate p63 expression status in giant cell tumor of bone in comparison with other giant cell-rich lesions. MATERIALS AND METHODS In a cross-sectional study we examined immunohistochemical expression of p63 in a series of 100 giant cell-rich bone lesions, including 31 giant cell tumors of bone, 14 osteosarcomas (including 3 giant cell-rich variants), 18 aneurysmal bone cysts (including one solid variant), 8 non-ossifying fibromas, 17 chondroblastomas, 8 tenosynovial giant cell tumors, and 4 brown tumors. RESULTS Immunohistochemical analysis showed p63 nuclear expression in 96.8% of giant cell tumors of bone, 14.3% of osteosarcomas, 50% of non-ossifiying fibromas, 22.2% of aneurysmal bone cysts, 68.7% of chondroblastomas, 75.0% of brown tumors and none of the tenosynovial giant cell tumors. Taking into account the intensity of staining, we identified strong staining in 48.4% of giant cell tumors of bone, 35.3% of chondroblastomas and 7.1% of osteosarcomas (in 2 cases which were both giant cell-rich variants). Considering extent of staining, extensive staining was only observed in 58.0% of giant cell tumors of bone, 23.5% of chondroblastomas and 14.3% of osteosarcomas. CONCLUSION A large number of giant cell tumors of bone (96.8%) are positive for p63, which is considerably more than any other giant cell-rich lesion. However, positive staining for p63 is not specific for GCTOB and may be seen in other lesions such as chondroblastoma, non-ossifying fibroma, brown tumor, and giant cell-rich osteosarcoma. P63 is a sensitive (96.8%) and relatively specific marker for discriminating GCTOB from other types of giant cell-rich lesions. We suggest a combined scoring method for p63 IHC staining interpretation in GC-rich lesions, considering both intensity and extent of reaction, with a 2+ cut off as a more accurate marker for the diagnosis of GCTOB within the appropriate clinical context.
Collapse
|
147
|
Abstract
Elucidating the mechanisms that regulate the life versus death of mammalian neurons is important not only for our understanding of the normal biology of the nervous system but also for our efforts to devise approaches to maintain neuronal survival in the face of traumatic injury or neurodegenerative disorders. Here, we review the emerging evidence that a key survival/death checkpoint in both peripheral and central neurons involves the p53 tumor suppressor and its newly discovered family members, p73 and p63. The full-length isoforms of these proteins function as proapoptotic proteins, whereas naturally occurring N-terminal truncated variants of p73 and p63 act as prosurvival proteins, at least partially by antagonizing the full-length family members. The authors propose that together, these isoforms comprise an upstream rheostat that sums different environmental cues to ultimately determine neuronal survival during development, during neuronal maintenance in adult animals, and even following traumatic injury.
Collapse
Affiliation(s)
- W Bradley Jacobs
- Developmental Biology and Cancer Research, Hospital for Sick Children, Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
148
|
Guo H, Yang S, Xu L, Li D, Tang J, Wang S, Wei B, Liu Z. Association between the p73 gene G4C14-to-A4T14 single nucleotide polymorphism and risk of cervical cancer by high resolution melting and PCR with confronting two-pair primers in a Chinese population. Oncol Lett 2016; 12:721-726. [PMID: 27347206 DOI: 10.3892/ol.2016.4655] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/18/2016] [Indexed: 11/06/2022] Open
Abstract
As a member of the p53 gene family, the p73 gene can affect an individual's susceptibility to cancer through a p53-like manner. DNA sequence variation in the p73 gene has been reported to be associated with cancer risk. The present study aimed to identify whether the p73 gene G4C14-to-A4T14 single nucleotide polymorphism (SNP) is associated with risk of cervical cancer in a Chinese population. The p73 G4C14-to-A4T14 polymorphism was genotyped in 175 cervical cancer and 189 healthy control peripheral blood DNA samples using high resolution melting, polymerase chain reaction with confronting two-pair primers and direct DNA sequencing. The results demonstrated that carriers of the AT/AT genotype were associated with a significantly increased risk of cervical cancer (P=0.042; χ2=4.122; odds ratio = 2.241; 95% confidence interval = 1.013-4.956) compared with the GC/GC genotype carriers. In addition, there was a significant association between p73 genotypes and tumor size in patients with cervical cancer (P=0.014; χ2=8.607). However, no association was identified between p73 genotypes and tumor stage, histological type or lymph node metastasis in patients with cervical cancer. These results suggest that the p73 G4C14-to-A4T14 SNP may function as a marker of genetic susceptibility to cervical cancer in the Chinese population.
Collapse
Affiliation(s)
- Haiyan Guo
- Department of Obstetrics and Gynecology, Xi'an No. 4 Hospital, Xi'an, Shaanxi 710004, P.R. China
| | - Shaodi Yang
- Institute of Biomedical Engineering, School of Geosciences and Info-Physics, Central South University, Changsha, Hunan 410000, P.R. China
| | - Lijian Xu
- Hunan Key Laboratory of Green Packaging and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, Hunan 412000, P.R. China
| | - Ding Li
- Hunan Key Laboratory of Green Packaging and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, Hunan 412000, P.R. China
| | - Jianxin Tang
- Hunan Key Laboratory of Green Packaging and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, Hunan 412000, P.R. China
| | - Shuangshaung Wang
- Hunan Key Laboratory of Green Packaging and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, Hunan 412000, P.R. China
| | - Benjie Wei
- Yin Feng Biological Group Co., Ltd., Jinan, Shandong 250000, P.R. China
| | - Zhengchun Liu
- Institute of Biomedical Engineering, School of Geosciences and Info-Physics, Central South University, Changsha, Hunan 410000, P.R. China
| |
Collapse
|
149
|
Nemajerova A, Kramer D, Siller SS, Herr C, Shomroni O, Pena T, Gallinas Suazo C, Glaser K, Wildung M, Steffen H, Sriraman A, Oberle F, Wienken M, Hennion M, Vidal R, Royen B, Alevra M, Schild D, Bals R, Dönitz J, Riedel D, Bonn S, Takemaru KI, Moll UM, Lizé M. TAp73 is a central transcriptional regulator of airway multiciliogenesis. Genes Dev 2016; 30:1300-12. [PMID: 27257214 PMCID: PMC4911929 DOI: 10.1101/gad.279836.116] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/02/2016] [Indexed: 01/25/2023]
Abstract
Motile multiciliated cells (MCCs) have critical roles in respiratory health and disease and are essential for cleaning inhaled pollutants and pathogens from airways. Despite their significance for human disease, the transcriptional control that governs multiciliogenesis remains poorly understood. Here we identify TP73, a p53 homolog, as governing the program for airway multiciliogenesis. Mice with TP73 deficiency suffer from chronic respiratory tract infections due to profound defects in ciliogenesis and complete loss of mucociliary clearance. Organotypic airway cultures pinpoint TAp73 as necessary and sufficient for basal body docking, axonemal extension, and motility during the differentiation of MCC progenitors. Mechanistically, cross-species genomic analyses and complete ciliary rescue of knockout MCCs identify TAp73 as the conserved central transcriptional integrator of multiciliogenesis. TAp73 directly activates the key regulators FoxJ1, Rfx2, Rfx3, and miR34bc plus nearly 50 structural and functional ciliary genes, some of which are associated with human ciliopathies. Our results position TAp73 as a novel central regulator of MCC differentiation.
Collapse
Affiliation(s)
- Alice Nemajerova
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Daniela Kramer
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Saul S Siller
- Department of Pharmacology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Christian Herr
- Department of Internal Medicine V, Saarland University, Homburg 66421, Germany
| | - Orr Shomroni
- Computational Systems Biology, German Center for Neurodegenerative Diseases, 37077 Göttingen, Germany
| | - Tonatiuh Pena
- Computational Systems Biology, German Center for Neurodegenerative Diseases, 37077 Göttingen, Germany
| | | | - Katharina Glaser
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Merit Wildung
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Henrik Steffen
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Anusha Sriraman
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Fabian Oberle
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Magdalena Wienken
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Magali Hennion
- Computational Systems Biology, German Center for Neurodegenerative Diseases, 37077 Göttingen, Germany
| | - Ramon Vidal
- Computational Systems Biology, German Center for Neurodegenerative Diseases, 37077 Göttingen, Germany
| | - Bettina Royen
- Department of Neurophysiology and Cellular Biophysics, Göttingen University, 37073 Göttingen, Germany
| | - Mihai Alevra
- Department of Neurophysiology and Cellular Biophysics, Göttingen University, 37073 Göttingen, Germany
| | - Detlev Schild
- Department of Neurophysiology and Cellular Biophysics, Göttingen University, 37073 Göttingen, Germany
| | - Robert Bals
- Department of Internal Medicine V, Saarland University, Homburg 66421, Germany
| | - Jürgen Dönitz
- Department of Evolutionary Developmental Genetics, Göttingen University, 37077 Göttingen, Germany
| | - Dietmar Riedel
- Electron Microscopy, Max-Planck-Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Stefan Bonn
- Computational Systems Biology, German Center for Neurodegenerative Diseases, 37077 Göttingen, Germany
| | - Ken-Ichi Takemaru
- Department of Pharmacology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, New York 11794, USA; Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany
| | - Muriel Lizé
- Institute of Molecular Oncology, Göttingen University, 37077 Göttingen, Germany; Clinic for Cardiology and Pneumology, Department of Pneumology, University Medical Center Göttingen, 37099 Göttingen, Germany
| |
Collapse
|
150
|
Solution structure and binding specificity of the p63 DNA binding domain. Sci Rep 2016; 6:26707. [PMID: 27225672 PMCID: PMC4880913 DOI: 10.1038/srep26707] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/09/2016] [Indexed: 01/17/2023] Open
Abstract
p63 is a close homologue of p53 and, together with p73, is grouped into the p53 family of transcription factors. p63 is known to be involved in the induction of controlled apoptosis important for differentiation processes, germ line integrity and development. Despite its high homology to p53, especially within the DNA binding domain (DBD), p63-DBD does not show cooperative DNA binding properties and is significantly more stable against thermal and chemical denaturation. Here, we determined the solution structure of p63-DBD and show that it is markedly less dynamic than p53-DBD. In addition, we also investigate the effect of a double salt bridge present in p53-DBD, but not in p63-DBD on the cooperative binding behavior and specificity to various DNA sites. Restoration of the salt bridges in p63-DBD by mutagenesis leads to enhanced binding affinity to p53-specific, but not p63-specific response elements. Furthermore, we show that p63-DBD is capable of binding to anti-apoptotic BclxL via its DNA binding interface, a feature that has only been shown for p53 so far. These data suggest that all p53 family members - despite alterations in the specificity and binding affinity - are capable of activating pro-apoptotic pathways in a tissue specific manner.
Collapse
|